• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 370
  • 208
  • 67
  • 49
  • 40
  • 29
  • 21
  • 11
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • Tagged with
  • 941
  • 179
  • 151
  • 122
  • 120
  • 102
  • 98
  • 91
  • 89
  • 76
  • 74
  • 74
  • 72
  • 71
  • 68
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
621

Defining the molecular role of RNA helicase DDX3 in antiviral signaling pathways / RNAヘリカーゼDDX3の抗ウイルス性シグナル伝達経路における分子的役割の解明

SAIKRUANG, WILAIPORN 23 May 2022 (has links)
京都大学 / 新制・課程博士 / 博士(生命科学) / 甲第24119号 / 生博第481号 / 新制||生||64(附属図書館) / 京都大学大学院生命科学研究科統合生命科学専攻 / (主査)教授 野田 岳志, 教授 鈴木 淳, 教授 高田 穣 / 学位規則第4条第1項該当 / Doctor of Philosophy in Life Sciences / Kyoto University / DFAM
622

Molecular Basis for Kappa-Opioid Regulation of Chemokine Receptor Function

Finley, Matthew James January 2009 (has links)
Opioid receptor-mediated regulation of chemokine receptors is vital for the host immune response, development, and neurological function. Previous studies have demonstrated that the kappa opioid receptor (KOR) activation results in decreased infectivity of human immunodeficiency virus 1 (HIV-1) in human peripheral blood mononuclear cells (PBMCs). We have found this effect is due to down-regulation of the major HIV-1 co-receptors, CCR5 and CXCR4. Using molecular techniques, CCR5 and CXCR4 mRNA levels drop dramatically following KOR activation. To dissect the mechanism involved, we used transcription factor binding arrays and compared control cell extracts to KOR activated cell extracts. We determined that the interferon regulatory factors (IRFs) and signal transducers and activators of transcription (STATs) could be involved in the KOR-mediated repression of CCR5 and CXCR4 transcription and protein expression. Using chemical inhibitors and small interfering RNA (siRNA) molecules, we determined that JAK2, STAT3, and IRF2 are critical members of this signal transduction pathway. The understanding of these particular mechanisms should prove to be beneficial for the development of potential pharmacological agents targeted at HIV-1 binding and infection since virus infection requires expression of the co-receptors CXCR4 and CCR5. Understanding the molecular basis for KOR-induced inhibition of co-receptor expression may provide a basis for the development of KOR agonist-based therapeutics to treat individuals infected with HIV. / Molecular Biology and Genetics
623

THE ROLE OF INTERFERON GAMMA AND CTLA4 IN MELANOCYTE AND MELANOMA BIOLOGY

Mo, Xuan January 2018 (has links)
Ultraviolet radiation (UVR) stimulates melanogenesis in melanocytes, primarily via release of alpha-melanocyte stimulating hormone from keratinocytes. UVR also induced an inflammatory response in the skin in which Interferon-gamma (IFNγ) cytokine plays an important orchestrating role. Here we report that recombinant IFNγ induces a temporal increase of melanogenesis in mouse melanoma cells. IFNγ elevates expression of microphthalmia-associated transcription factor (Mitf), which is the master regulator of melanogenesis by initiating transcription of melanogenic enzymes, tyrosinase (Tyr), tyrosinese-related protein 1 (Tyrp1) and dopachrome tautomerase (Dct). Interestingly, tyrosinase protein, but not mRNA expression, accumulated in response to IFNγ treatment and was consistent with tyrosinase activity. In addition, glycosidase digestion showed that IFNγ induced ER-resistant, fully mature tyrosinase via post-transcriptional mechanisms, rather than increased de novo synthesis or early processing in the ER. Most strikingly, IFNγ mediated alkalization of melanosomes by elevating Oca2 expression, which leads to facilitate melanosome maturation and sequential accumulation of mature tyrosianse. Both Jak1/Jak2 inhibitor Ruxolitinib and knockout of Stat1 mediated by CRISPR-CAS9 blocked the IFNγ-induced Mitf, tyrosinase, Oca2 expressions and melanin biosynthesis. Our data reveals that IFNγ-Jak1/2-Stat1 axis regulates melanogenesis by inducing maturation of melanosomes and accumulation of mature tyrosinase via post-translational mechanisms. CTLA4 is a cell surface receptor on T cells that functions as an immune checkpoint molecule to enforce tolerance to cognate antigens. Anti-CTLA4 immunotherapy is highly effective at reactivating T cell responses against melanoma, which is postulated to be due to targeting CTLA4 on T cells. Here we report that CTLA4 is also highly expressed by most human melanoma cell lines, as well as in normal human melanocytes. Interferon-gamma (IFNγ) signaling activated the expression of the human CTLA4 gene in a melanocyte and melanoma cell-specific manner. Mechanistically, IFNγ activated CTLA4 expression through JAK1/2-dependent phosphorylation of STAT1, which bound a specific gamma-activated sequence (GAS) site on the CTLA4 promoter, thereby licensing CBP/p300-mediated histone acetylation and local chromatin opening. In melanoma cell lines, elevated baseline expression relied upon constitutive activation of the MAPK pathway. Notably, RNA-seq analyses of melanoma specimens obtained from patients who had received anti-CTLA4 immunotherapy (ipilimumab) showed upregulation of an IFNγ -response gene expression signature, including CTLA4 itself, which correlated significantly with durable response. We also show that ectopic expression of Ctla4 in mouse melanoma cells promotes tumor growth in immunocompetent mice. Ctla4-enhanced melanomagenesis is blocked in immunodeficient NSG mice. In addition, ligation of CD86 (one of Ctla4 ligands) in T cells inhibits CD8 T cells proliferation in vitro. Expression of Ctla4 in melanoma cells are resistant to CD8 T cell cytoxicity in vitro. Our data demonstrates and highlights the novel and unrecognized functions of CTLA4 in melanoma cells that aids their survival, immunoevasion and tumorigenic capabilities. Taken together, these findings have potential implications for the conventional and prototypical roles of the IFNγ signaling pathway and CTLA4 in tumor immunosurveillance and tumor immunoevasion. More importantly, our results raise the possibility that CTLA4 targeting on melanoma cells may contribute to the clinical immunobiology of anti-CTLA4 responses. / Biomedical Sciences
624

Interferon-gamma and the regulation of neuroinflammation

Millward, Jason Michael, 1976- January 2008 (has links)
No description available.
625

Influenza A Virus PB1-F2 Protein: its Role in Pathogenesis

Deventhiran, Jagadeeswaran 31 July 2015 (has links)
Influenza A virus (IAV) causes annual seasonal epidemics and occasional pandemics resulting in significant levels of mortality and socio-economic costs worldwide. PB1-F2 is a small non-structural protein encoded by an alternate +1 open reading frame in the PB1 gene. PB1-F2 is considered to play important roles in primary influenza virus infection and post-influenza secondary bacterial pneumonia in mice. It is a multifunctional and enigmatic protein with diverse functions attributed to it and the precise contribution of PB1-F2 to the IAV life cycle in avian and mammalian hosts remains largely unknown. In the triple-reassortant H3N2 (TR H3N2) swine influenza virus (SIV) background, we found that PB1-F2 expression did not affect nasal shedding, lung viral load, immunophenotypes, and lung pathology in pigs. On the other hand, in turkeys, deletion of PB1-F2 resulted in early induction of clinical disease and effective transmission among the turkey poults. Interestingly, the virulence associated 66S mutation in PB1-F2 abolished the ability of the IAV to successfully infect turkeys and transmit to in-contacts. These results highlight the strain- and species-specific role of PB1-F2 protein. We also demonstrated that specific amino acid residues in the C-terminal of PB1-F2 determine the pathogenicity of 2009 swine-origin pandemic H1N1 virus in a mouse model. The C-terminal residues 73K, 75R, and 79R together with 66S increased virus replication, decreased type I interferon response, increased infiltration of neutrophils and myeloperoxidase production in lungs resulting in acute respiratory distress syndrome (ARDS) in mice with characteristic clinical and pathological features of acute lung injury (ALI). Further, we found that PB1-F2 induces mitochondrial superoxide production and mitochondrial damage in a sequence dependent manner in IAV-infected lung epithelial cells. PB1-F2-mediated mitochondrial damage promotes Parkin-mediated mitophagy but suppresses the autophagic degradation of damaged mitochondria in the infected lung epithelial cells. Accumulated dysfunctional mitochondria likely to aggravate host cell death and inflammatory responses. Taken together, the present findings enhance our understanding of PB1-F2 protein as a virulence determinant in IAV infection in a species- and strain-specific manner and provide new insights into the impact of genetic changes in PB1-F2 on the host pathogenesis of virulent IAV strains. / Ph. D.
626

Tissue factor binds to and inhibits interferon-alpha receptor-1 signaling

Manoharan, Jayakumar 16 September 2024 (has links)
No description available.
627

IFNλ stimulates MxA production in human dermal fibroblasts via a MAPK-dependent STAT1-independent mechanism

Alase, Adewonuola A., El-Sherbiny, Y., Vital, E., Tobin, Desmond J., Turner, N.A., Wittmann, Miriam 08 1900 (has links)
Yes / Interferon lambda (IFNλ) is important for epidermal defence against viruses. It is produced by, and acts on, keratinocytes, whereas fibroblasts were previously considered to be unresponsive to this type III IFN. Herein we report findings revealing cell type-specific differences in IFNλ signalling and function in skin resident cells. In dermal fibroblasts, IFNλ induced the expression of MxA, a potent antiviral factor, but not other IFN signature genes as it does in primary keratinocytes. In contrast to its effect on keratinocytes, IFNλ did not phosphorylate STAT1 in fibroblasts, but instead activated MAPKs. Accordingly, inhibition of MAPK activation (p38 and p42/44) blocked the expression of MxA protein in fibroblasts but not in keratinocytes. Functionally, IFNλ inhibited proliferation in keratinocytes but not in fibroblasts. Moreover, IFNλ upregulated the expression of TGFβ1-induced collagens in fibroblasts. Taken together, our findings identify primary human dermal fibroblasts as responder cells to IFNλ. Our study shows cutaneous cell type-specific IFN signalling and suggests that IFNλ, whilst important for epidermal anti-viral competence, may also have a regulatory role in the dermal compartment balancing type I IFN-induced inhibition of tissue repair processes.
628

Aryl hydrocarbon receptor expression in lung adenocarcinoma promotes immunosuppression in the tumor microenvironment

Snyder, Megan Sara 30 October 2024 (has links)
The discovery of immune checkpoints and the subsequent development of pharmacological immune checkpoint inhibitors generated excitement and optimism that researchers had finally found the key to inducing complete remission of tumors. Inhibitors targeting the PD-1/PD-L1 signaling axis, though still facing limitations with widespread utility, have proven to be beneficial in multiple types of cancerous tissues. While some patients experience dramatic results, immune checkpoint inhibitors are not the panacea that was initially hoped. Why anti-PD-1/anti-PD-L1 therapies often come up short might be that the underlying regulators of PD-L1 expression or other molecular regulators of immune suppression are not being addressed by the pharmacological immunomodulators. One protein often active in the tumor microenvironment (TME) is involved in both these possibilities: the aryl hydrocarbon receptor (AhR). The AhR is a ligand-activated transcription factor that can suppress immune activation as well as drive cancer progression by regulating transcription of a myriad of different genes within both malignant and immune cells. While AhR activation often correlates with poor prognosis in a variety of cancers, how AhR activation affects functional characteristics within and between malignant cells and immune cells in the TME is not fully understood. This is especially true in lung cancer, one of the most common and deadliest malignancies, which develops in an organ with many tissue-resident immune cells and opportunities to encounter exogenous pathogens—and environmental AhR agonists. To this end, we sought to elucidate the interactions between the AhR and its immunomodulatory target genes, PD-L1 and IDO1, in malignant lung adenocarcinoma (LUAD) cells and to determine how these interactions influence immunosuppression in the greater TME. The following work describes how AhR activation in LUAD cells results in the upregulation of two immune checkpoints, PD-L1 and IDO, in the malignant cell. We showed that AhR expression in CMT167 LUAD cells led to rapid tumor growth following subcutaneous or orthotopic transplantation whereas AhR deletion resulted in slower proliferation and up to complete rejection of tumor cells. AhR knockout also resulted in greater infiltration of CD3+ T cells. T cells from CMT167AhR-KO tumors were characterized by expression of granzyme B and upregulation of genes involved in activated and cytotoxic immune response, while T cells from CMT167WT tumors expressed higher levels of genes associated with T cell exhaustion and non-Th1 phenotypes. In addition, we developed a novel orthotopic model that will allow us to better understand how immune checkpoints from tumor cells modulate the lung TME by tracing the interactions of both myeloid and lymphoid immune cells with malignant cells. Further, in human LUAD cells, we implicated the AhR in the control of a long non-coding RNA shown to promote PD-L1 expression in the presence of IFNγ. Together, these results implicate AhR as a regulator of immune suppression in LUAD through multiple mechanisms and support it as a therapeutic target in the quest to improve immunotherapies.
629

Avaliação do papel da imunidade adaptativa na obesidade: estudo experimental em animais / Evaluation of the role of adaptative immunity in obesity: study in animals

Giraldez, Viviane Zorzanelli Rocha 23 July 2014 (has links)
O desenvolvimento gradual e recente de uma epidemia mundial de obesidade alavancou sobremaneira o estudo dessa condição e de suas comorbidades metabólicas. No âmbito fisiopatológico, múltiplos estudos demonstraram a expressão aumentada de mediadores inflamatórios no tecido adiposo de animais e humanos obesos, o acúmulo local de macrófagos, e um papel central da inflamação no desequilíbrio da homeostase metabólica local e sistêmica na obesidade. A definição de um papel ativo dos macrófagos, e portanto da imunidade inata, na rede inflamatória do tecido adiposo, evocou a hipótese de que, similarmente a outras condições inflamatórias crônicas como a aterosclerose, a obesidade também contaria com a importante participação de elementos da imunidade adaptativa, como as células T e suas citocinas, em sua fisiopatologia. Com base nessas considerações, os objetivos principais desse estudo foram: 1) avaliar a presença das células T e o papel do interferon-gama (IFNy), clássica citocina T-helper 1 (ou Th1), na inflamação do tecido adiposo; e 2) estudar mecanismos de acúmulo das células T no tecido adiposo na obesidade, particularmente a participação do receptor CXCR3 nesse processo. Experimentos de citometria de fluxo mostraram que o tecido adiposo visceral de camundongos C57BL/6 obesos após consumo de dieta rica em gorduras apresentou maior número de macrófagos e também de células T, CD4+ e CD8+, em comparação a controles que receberam dieta pobre em gorduras. A expressão de I-Ab, marcador do complexo de histocompatibilidade principal classe II (MHC II) murino, também foi maior no tecido adiposo dos animais obesos, sugerindo a presença local da atividade de apresentação de antígeno com consequente ativação das células T. Quando estimuladas in vitro, células T derivadas do tecido adiposo de camundongos obesos produziram mais IFNy do que aquelas isoladas de controles, novamente sugerindo a ativação dessas células em um contexto de obesidade. Na análise das possíveis funções do IFNy no tecido adiposo, a estimulação da linhagem de células 3T3-L1 diferenciadas em adipócitos com IFNy recombinante resultou na produção aumentada de quimiocinas de macrófagos, como a proteína quimiotática de monócito (MCP-1), e de quimiocinas de células T, como a proteína 10 induzida por IFNy (IP-10) e monocina induzida por IFNy (MIG). A estimulação de adipócitos com o sobrenadante de células Th1 cultivadas in vitro, com abundante concentração de IFNy, também levou à produção aumentada de IP-10. Em análise mais ampla, através de microarray, dos possíveis efeitos do IFNy na expressão gênica de adipócitos, o tratamento dessas células com 100 U/ml de IFNy resultou na expressão aumentada de diversas quimiocinas e seus receptores em comparação ao grupo tratado com placebo. Similarmente à estimulação de células isoladas com IFNy, a incubação de tecido adiposo ex vivo de camundongos com essa citocina também resultou em secreção aumentada de IP-10, MIG e fator de necrose tumoral alfa (TNFy). A investigação do papel do IFNy na inflamação do tecido adiposo in vivo envolveu camundongos com deficiência de IFNy e controles, ambos os grupos submetidos a dieta rica em gorduras (obesos) ou pobre em gorduras (não obesos). Camundongos obesos deficientes em IFNy apresentaram expressão reduzida de mRNA de genes inflamatórios como TNFalfa e MCP-1 no tecido adiposo; acúmulo local reduzido de macrófagos; e melhor tolerância à glicose em comparação aos controles sob mesma dieta. Animais com deficiência de apolipoproteína E (ApoE) e também do receptor de IFNy também apresentaram em seu tecido adiposo a expressão reduzida de mRNA de genes inflamatórios, particularmente relacionados às células T, como IP-10, MIG, e o receptor CXCR3, em comparação aos controles com deficiência única de ApoE. Resultados in vitro e in vivo sugerem conjuntamente um importante papel do IFNy, e portanto, das células T e da imunidade adaptativa, na rede inflamatória do tecido adiposo na obesidade, com consequente impacto metabólico sistêmico. A presença de células T ativadas no tecido adiposo e seu acúmulo diferencial na obesidade motivaram também a pesquisa de potenciais mecanismos quimiotáticos reguladores desse processo. CXCR3, receptor das quimiocinas de células T, IP-10, MIG e quimiocina alfa de células T IFNy-induzida (I-TAC), é expresso preferencialmente em células T ativadas, e detém papel central na migração dessas células em outras condições inflamatórias crônicas, como a aterosclerose. Em camundongos com deficiência de CXCR3 e que receberam dieta rica em gorduras por 8 ou 16 semanas, o tecido adiposo apresentou significativamente menos células T, incluindo as células CD4+ e CD8+, em comparação a controles submetidos a mesma dieta. Os números similares de células T e outras populações de leucócitos no baço e sangue periférico dos animais deficientes em CXCR3 e controles fortalecem o conceito de um efeito do CXCR3 sobre o acúmulo de células T no tecido adiposo, independentemente do número de células circulantes e periféricas. Os camundongos deficientes em CXCR3 apresentaram também maior tolerância à glicose e expressão reduzida de mRNA de mediadores inflamatórios em seu tecido adiposo em comparação aos controles após 8 semanas de dieta rica em gorduras. No entanto, a diferença na tolerância à glicose entre os dois grupos tornou-se não significativa após 16 semanas de dieta gordurosa, coincidindo com redução substancial na expressão de mRNA de mediadores anti-inflamatórios (como interleucina-10 [IL-10] e Arginase 1), e número reduzido de células T regulatórias no tecido adiposo de camundongo s deficientes em CXCR3 em relação a controles. Esses resultados sugerem que o CXCR3 é capaz de regular o acúmulo de células T de diferentes subtipos, com perfil proinflamatório ou anti-inflamatório. Em conclusão, nossos resultados revelam um importante papel da citocina Th1 IFNy na rede inflamatória do tecido adiposo na obesidade em camundongos, sugerindo a participação fundamental das células T e portanto, da imunidade adaptativa nesse cenário. Além disso, o receptor CXCR3 contribui significativamente para o acúmulo das células T, incluindo as células T regulatórias, no tecido adiposo desses animais / The gradual and recent development of a worldwide epidemic of obesity greatly leveraged the study of this condition and its metabolic comorbidities. In the pathophysiologic context, multiple studies have demonstrated increased expression of inflammatory mediators in adipose tissue of obese animals and humans, the local macrophage accumulation, and a central role of inflammation in the imbalance of local or systemic metabolic homeostasis in obesity. The concept of an active role of macrophages and thus of innate immunity in the inflammatory network of adipose tissue, suggested the hypothesis that, similar to other chronic inflammatory conditions such as atherosclerosis, obesity also count on the participation of important elements of adaptive immunity such as T cells and their cytokines in its pathophysiology. Based on these considerations, the main objectives of this study were: 1) to evaluate the presence of T cells and the role of interferon-gamma (IFNy), classic T-helper 1 (Th1) cytokine, in adipose tissue inflammation, and 2) to study mechanisms of T cell accumulation in adipose tissue in the context of obesity, particularly the involvement of CXCR3 receptor in this process. Flow cytometry experiments showed that the visceral fat tissue of C57BL/6 obese mice fed a high fat diet showed a greater number of macrophages and also T cells, including CD4+ and CD8+ cells, compared to controls fed a low-fat diet. The expression of I-Ab, murine marker of class II major histocompatibility complex (MHC II), was also higher in adipose tissue of obese animals, suggesting the presence of local antigen presentation and consequent T cell activation. When stimulated in vitro, T cells derived from adipose tissue of obese mice produced more IFNy than those isolated from controls, again suggesting the activation of these cells in the context of obesity. In the analysis of possible functions of IFNy in adipose tissue, stimulation of 3T3 -L1 cells differentiated into adipocytes with recombinant IFNy resulted in enhanced production of macrophage chemokines, such as monocyte chemotactic protein-1 (MCP-1) and T-cell chemokines, such as interferon gamma-induced protein 10 (IP-10) and monokine induced by gamma interferon (MIG). The stimulation of adipocytes with the supernatant of in vitro cultured Th1 cells, with abundant levels of IFNy, has also led to increased IP-10 production. In a broader analysis, by microarray, of the possible effects of IFNy on adipocyte gene expression, treatment of these cells with 100 U/ml of IFNy resulted in increased expression of chemokines and their receptors in comparison to the placebo group. Similarly to the stimulation of isolated cells with IFNy, incubation of ex vivo adipose tissue with this cytokine also resulted in increased IP-10, MIG and tumor necrosis factor alpha (TNFalpha) secretion
630

Investigação da resposta imunológica antitumoral induzida por células B16F10 tratadas pela combinação p19Arf e interferon-beta em um modelo de vacinação profilático para melanoma murino / Investigation of the antitumor immune response induced by B16F10 cells treated with the p19Arf and Interferon-beta combination in a murine prophylatic model of melanoma vaccine

Medrano, Ruan Felipe Vieira 25 April 2013 (has links)
Dados recentes do nosso laboratório demonstram que somente a co-transdução, não a tradução individual, com vetores adenovirais portadores de Interferon-beta (IFN?) (citocina imuno modulatória) e p19Arf (parceira funcional da proteína supressora de tumor p53) resulta na morte celular massiva do melanoma murino B16F10. A capacidade desse tratamento combinado de induzir uma resposta imune antitumoral ainda não foi avaliada. Dessa maneira, o objetivo do presente trabalho foi investigar se células B16F0 tratadas por essa combinação são capazes de induzir uma resposta imune antitumoral em um modelo de vacinação profilático de melanoma. Para isso, essas células foram co-transduzidas com os vetores AdPGp19 e AdPGIFN? e 48 horas depois, inoculadas como agente vacinal no flanco esquerdo (sítio da vacina) de camundongos C57BL/6 imunocompetentes. Sete dias após a última vacinação, esses animais foram desafiados com células B16F10 naïve no flanco direito (sítio do desafio). A progressão tumoral do desafio foi significativamente reduzida, mesmo quando o desafio tumoral foi feito 73 dias após da vacinação. Porém, como os animais imunizados desenvolveram tumores no sítio da vacina, condições para o uso dessas células tratadas foram avaliadas, revelando que: o número de células e de aplicações usadas durante a vacinação tem influência no aparecimento desse tumores, e que apenas com o tratamento combinado os camundongos permanecem livres de tumor. A influência do sistema imune para este resultado foi revelada após protocolo de imunussupressão. Em seguida, o papel da p19Arf e do IFN? na proteção antitumoral da combinação foi estudado. In vitro, os efeitos antitumorais da combinação parecem ser mais influentes da reposição de p19Arf do que da expressão de IFN?, mas já in vivo, na presença do sistema imune, foram mais dependentes do IFN?. Com a combinação estes efeitos mostraram-se mais pronunciados, induzindo uma proteção antitumoral e maior sobrevida aos animais vacinados. Estes resultados indicam que a combinação p19Arf e IFN? pode ser aplicada como um agente imunoterápico e sugerem que a associação entre morte celular e imuno estimulação pode beneficiar o tratamento contra o câncer / Previously, we have shown in a mouse melanoma model of in situ gene therapy that co-transduction, but not individual application, with adenovirus vectors expressing the Interferon-beta (IFN?) (immune modulatory cytokine) and p19Arf (functional partner of the p53 tumor suppressor) transgenes results in massive cell death and reduced tumor progression. However, the capability of this combined treatment to stimulate an antitumor immune response has not been evaluated. Therefore, the aim of this work was to investigate, trough a prophylactic vaccine model, if B16F10 cells treated by the p19Arf and IFN? combination could induce such immune response. To do so, these cells were co-transduced by the AdPGp19 e AdPGIFN? adenoviral vectors and 48 hours after, inoculated as a vaccine agent in the left flank (vaccine site) of immune competent C57BL/6 mice. Seven days after the last vaccine, a tumor challenge was done with naïve B16F10 cells in the right flank (challenge site). Tumor progression was markedly reduced, even when challenge was done 73 days after the vaccination. However, since these animals developed tumors where the vaccine was applied, more appropriate conditions for the use of these treated cells were pursued, thus revealing that: the number of cells and inoculations can dictate tumor development, and also, that only with the combined treatment was tumor formation abolished. The influence of the immune system for this result was revelead by performing an immune supression protocol. Next, the roles of p19Arf and of IFN? were studied. In vitro, the antitumor effects were stronger upon the introduction of p19Arf than IFN?, but in vivo, in the presence of the immune system, the effects were more IFN? dependent. In fact, these effects were more pronouced with the combined treatment, inducing protection against tumor formation and progression and increasing survival in the vaccinated animals. Taken together, these results demonstrate the application of cells treated by the p19Arf e IFN? combination as an effective vaccine agent and also indicates that the association between cell death and immune stimulation may benefit the treatment of cancer

Page generated in 0.1844 seconds